Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cell ; 187(1): 110-129.e31, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38181737

ABSTRACT

X chromosome inactivation (XCI) serves as a paradigm for RNA-mediated regulation of gene expression, wherein the long non-coding RNA XIST spreads across the X chromosome in cis to mediate gene silencing chromosome-wide. In female naive human pluripotent stem cells (hPSCs), XIST is in a dispersed configuration, and XCI does not occur, raising questions about XIST's function. We found that XIST spreads across the X chromosome and induces dampening of X-linked gene expression in naive hPSCs. Surprisingly, XIST also targets specific autosomal regions, where it induces repressive chromatin changes and gene expression dampening. Thereby, XIST equalizes X-linked gene dosage between male and female cells while inducing differences in autosomes. The dispersed Xist configuration and autosomal localization also occur transiently during XCI initiation in mouse PSCs. Together, our study identifies XIST as the regulator of X chromosome dampening, uncovers an evolutionarily conserved trans-acting role of XIST/Xist, and reveals a correlation between XIST/Xist dispersal and autosomal targeting.


Subject(s)
Genes, X-Linked , RNA, Long Noncoding , X Chromosome , Animals , Female , Humans , Male , Mice , Gene Silencing , RNA, Long Noncoding/genetics , X Chromosome/genetics , Pluripotent Stem Cells/metabolism
2.
Nucleic Acids Res ; 51(5): 2177-2194, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36727460

ABSTRACT

X chromosome inactivation (XCI) is an essential process, yet it initiates with remarkable diversity in various mammalian species. XIST, the main trigger of XCI, is controlled in the mouse by an interplay of lncRNA genes (LRGs), some of which evolved concomitantly to XIST and have orthologues across all placental mammals. Here, we addressed the functional conservation of human orthologues of two such LRGs, FTX and JPX. By combining analysis of single-cell RNA-seq data from early human embryogenesis with various functional assays in matched human and mouse pluripotent stem- or differentiated post-XCI cells, we demonstrate major functional differences for these orthologues between species, independently of primary sequence conservation. While the function of FTX is not conserved in humans, JPX stands as a major regulator of XIST expression in both species. However, we show that different entities of JPX control the production of XIST at various steps depending on the species. Altogether, our study highlights the functional versatility of LRGs across evolution, and reveals that functional conservation of orthologous LRGs may involve diversified mechanisms of action. These findings represent a striking example of how the evolvability of LRGs can provide adaptative flexibility to constrained gene regulatory networks.


Subject(s)
Placenta , RNA, Long Noncoding , Pregnancy , Humans , Female , Mice , Animals , Placenta/metabolism , X Chromosome Inactivation/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mammals/genetics , Embryo, Mammalian/metabolism
4.
Stem Cell Reports ; 17(10): 2220-2238, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36179695

ABSTRACT

Telencephalic organoids generated from human pluripotent stem cells (hPSCs) are a promising system for studying the distinct features of the developing human brain and the underlying causes of many neurological disorders. While organoid technology is steadily advancing, many challenges remain, including potential batch-to-batch and cell-line-to-cell-line variability, and structural inconsistency. Here, we demonstrate that a major contributor to cortical organoid quality is the way hPSCs are maintained prior to differentiation. Optimal results were achieved using particular fibroblast-feeder-supported hPSCs rather than feeder-independent cells, differences that were reflected in their transcriptomic states at the outset. Feeder-supported hPSCs displayed activation of diverse transforming growth factor ß (TGFß) superfamily signaling pathways and increased expression of genes connected to naive pluripotency. We further identified combinations of TGFß-related growth factors that are necessary and together sufficient to impart broad telencephalic organoid competency to feeder-free hPSCs and enhance the formation of well-structured brain tissues suitable for disease modeling.


Subject(s)
Organoids , Pluripotent Stem Cells , Cell Differentiation/physiology , Humans , Organoids/metabolism , Pluripotent Stem Cells/metabolism , Telencephalon/metabolism , Transforming Growth Factor beta/metabolism
5.
Nat Cell Biol ; 24(6): 858-871, 2022 06.
Article in English | MEDLINE | ID: mdl-35697783

ABSTRACT

Human naive pluripotent stem cells have unrestricted lineage potential. Underpinning this property, naive cells are thought to lack chromatin-based lineage barriers. However, this assumption has not been tested. Here we define the chromatin-associated proteome, histone post-translational modifications and transcriptome of human naive and primed pluripotent stem cells. Our integrated analysis reveals differences in the relative abundance and activities of distinct chromatin modules. We identify a strong enrichment of polycomb repressive complex 2 (PRC2)-associated H3K27me3 in the chromatin of naive pluripotent stem cells and H3K27me3 enrichment at promoters of lineage-determining genes, including trophoblast regulators. PRC2 activity acts as a chromatin barrier restricting the differentiation of naive cells towards the trophoblast lineage, whereas inhibition of PRC2 promotes trophoblast-fate induction and cavity formation in human blastoids. Together, our results establish that human naive pluripotent stem cells are not epigenetically unrestricted, but instead possess chromatin mechanisms that oppose the induction of alternative cell fates.


Subject(s)
Pluripotent Stem Cells , Polycomb Repressive Complex 2 , Cell Differentiation/genetics , Chromatin/genetics , Histones/genetics , Humans , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Trophoblasts/metabolism
6.
Sci Adv ; 8(12): eabk0013, 2022 03 25.
Article in English | MEDLINE | ID: mdl-35333572

ABSTRACT

Uncovering the mechanisms that establish naïve pluripotency in humans is crucial for the future applications of pluripotent stem cells including the production of human blastoids. However, the regulatory pathways that control the establishment of naïve pluripotency by reprogramming are largely unknown. Here, we use genome-wide screening to identify essential regulators as well as major impediments of human primed to naïve pluripotent stem cell reprogramming. We discover that factors essential for cell state change do not typically undergo changes at the level of gene expression but rather are repurposed with new functions. Mechanistically, we establish that the variant Polycomb complex PRC1.3 and PRDM14 jointly repress developmental and gene regulatory factors to ensure naïve cell reprogramming. In addition, small-molecule inhibitors of reprogramming impediments improve naïve cell reprogramming beyond current methods. Collectively, this work defines the principles controlling the establishment of human naïve pluripotency and also provides new insights into mechanisms that destabilize and reconfigure cell identity during cell state transitions.


Subject(s)
Cellular Reprogramming , Pluripotent Stem Cells , Polycomb Repressive Complex 1 , Cell Differentiation , Gene Expression Regulation , Humans , Pluripotent Stem Cells/cytology , Polycomb Repressive Complex 1/metabolism
7.
Elife ; 102021 08 31.
Article in English | MEDLINE | ID: mdl-34463252

ABSTRACT

The signalling pathways that maintain primed human pluripotent stem cells (hPSCs) have been well characterised, revealing a critical role for TGFß/Activin/Nodal signalling. In contrast, the signalling requirements of naive human pluripotency have not been fully established. Here, we demonstrate that TGFß signalling is required to maintain naive hPSCs. The downstream effector proteins - SMAD2/3 - bind common sites in naive and primed hPSCs, including shared pluripotency genes. In naive hPSCs, SMAD2/3 additionally bind to active regulatory regions near to naive pluripotency genes. Inhibiting TGFß signalling in naive hPSCs causes the downregulation of SMAD2/3-target genes and pluripotency exit. Single-cell analyses reveal that naive and primed hPSCs follow different transcriptional trajectories after inhibition of TGFß signalling. Primed hPSCs differentiate into neuroectoderm cells, whereas naive hPSCs transition into trophectoderm. These results establish that there is a continuum for TGFß pathway function in human pluripotency spanning a developmental window from naive to primed states.


Subject(s)
Cell Differentiation/physiology , Pluripotent Stem Cells/physiology , Signal Transduction/physiology , Smad2 Protein/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Cell Line , Cellular Reprogramming , Humans , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism
8.
Curr Opin Genet Dev ; 70: 89-96, 2021 10.
Article in English | MEDLINE | ID: mdl-34246082

ABSTRACT

Transcription factor (TF)-induced reprogramming of somatic cells across lineages and to induced pluripotent stem cells (iPSCs) has revealed a remarkable plasticity of differentiated cells and presents great opportunities for generating clinically relevant cell types for disease modeling and regenerative medicine. The understanding of iPSC reprogramming provides insights into the mechanisms that safeguard somatic cell identity, drive epigenetic reprogramming, and underlie cell fate specification in vivo. The combinatorial action of TFs has emerged as the key mechanism for the direct and indirect effects of reprogramming factors that induce the remodelling of the enhancer landscape. The interplay of TFs in iPSC reprogramming also yields trophectoderm- and extraembryonic endoderm-like cell populations, uncovering an intriguing plasticity of cell states and opening new avenues for exploring cell fate decisions during early embryogenesis.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/chemistry , Transcription Factors/metabolism , Animals , Embryonic Development , Epigenesis, Genetic , Humans , Induced Pluripotent Stem Cells/metabolism
9.
Nat Commun ; 12(1): 2098, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33828098

ABSTRACT

The transition from naive to primed pluripotency is accompanied by an extensive reorganisation of transcriptional and epigenetic programmes. However, the role of transcriptional enhancers and three-dimensional chromatin organisation in coordinating these developmental programmes remains incompletely understood. Here, we generate a high-resolution atlas of gene regulatory interactions, chromatin profiles and transcription factor occupancy in naive and primed human pluripotent stem cells, and develop a network-graph approach to examine the atlas at multiple spatial scales. We uncover highly connected promoter hubs that change substantially in interaction frequency and in transcriptional co-regulation between pluripotent states. Small hubs frequently merge to form larger networks in primed cells, often linked by newly-formed Polycomb-associated interactions. We identify widespread state-specific differences in enhancer activity and interactivity that correspond with an extensive reconfiguration of OCT4, SOX2 and NANOG binding and target gene expression. These findings provide multilayered insights into the chromatin-based gene regulatory control of human pluripotent states.


Subject(s)
Gene Expression Regulation , Pluripotent Stem Cells/metabolism , Chromatin/metabolism , DNA Methylation , Enhancer Elements, Genetic , Humans , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Promoter Regions, Genetic , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors/metabolism
10.
Cell Rep ; 33(13): 108562, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33378680

ABSTRACT

Generating mammalian cells with desired mitochondrial DNA (mtDNA) sequences is enabling for studies of mitochondria, disease modeling, and potential regenerative therapies. MitoPunch, a high-throughput mitochondrial transfer device, produces cells with specific mtDNA-nuclear DNA (nDNA) combinations by transferring isolated mitochondria from mouse or human cells into primary or immortal mtDNA-deficient (ρ0) cells. Stable isolated mitochondrial recipient (SIMR) cells isolated in restrictive media permanently retain donor mtDNA and reacquire respiration. However, SIMR fibroblasts maintain a ρ0-like cell metabolome and transcriptome despite growth in restrictive media. We reprogrammed non-immortal SIMR fibroblasts into induced pluripotent stem cells (iPSCs) with subsequent differentiation into diverse functional cell types, including mesenchymal stem cells (MSCs), adipocytes, osteoblasts, and chondrocytes. Remarkably, after reprogramming and differentiation, SIMR fibroblasts molecularly and phenotypically resemble unmanipulated control fibroblasts carried through the same protocol. Thus, our MitoPunch "pipeline" enables the production of SIMR cells with unique mtDNA-nDNA combinations for additional studies and applications in multiple cell types.


Subject(s)
Cellular Reprogramming , Fibroblasts/metabolism , Gene Transfer Techniques , High-Throughput Screening Assays/methods , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/transplantation , Animals , Cell Differentiation , Cell Line , DNA, Mitochondrial/metabolism , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Metabolome , Mice , Mice, Inbred C57BL , Transcriptome
11.
Stem Cell Reports ; 14(5): 972-988, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32302559

ABSTRACT

Naive and primed human pluripotent stem cells (hPSC) provide valuable models to study cellular and molecular developmental processes. The lack of detailed information about cell-surface protein expression in these two pluripotent cell types prevents an understanding of how the cells communicate and interact with their microenvironments. Here, we used plasma membrane profiling to directly measure cell-surface protein expression in naive and primed hPSC. This unbiased approach quantified over 1,700 plasma membrane proteins, including those involved in cell adhesion, signaling, and cell interactions. Notably, multiple cytokine receptors upstream of JAK-STAT signaling were more abundant in naive hPSC. In addition, functional experiments showed that FOLR1 and SUSD2 proteins are highly expressed at the cell surface in naive hPSC but are not required to establish human naive pluripotency. This study provides a comprehensive stem cell proteomic resource that uncovers differences in signaling pathway activity and has identified new markers to define human pluripotent states.


Subject(s)
Cell Adhesion , Cell Membrane/metabolism , Induced Pluripotent Stem Cells/metabolism , Proteome/genetics , Signal Transduction , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Folate Receptor 1/genetics , Folate Receptor 1/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Proteome/metabolism , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism
12.
Bioessays ; 40(5): e1700239, 2018 05.
Article in English | MEDLINE | ID: mdl-29574793

ABSTRACT

Recent reports that human pluripotent stem cells can be captured in a spectrum of states with variable properties has prompted a re-evaluation of how pluripotency is acquired and stabilised. The latest additions to the stem cell hierarchy open up opportunities for understanding human development, reprogramming, and cell state transitions more generally. Many of the new cell lines have been collectively termed 'naïve' human pluripotent stem cells to distinguish them from the conventional 'primed' cells. Here, several transcriptional and epigenetic hallmarks of human pluripotent states in the recently described cell lines are reviewed and evaluated. Methods to derive and identify human naïve pluripotent stem cells are also discussed, with a focus on the uses and future developments of state-specific reporter cell lines and cell-surface proteins. Finally, opportunities and uncertainties in naïve stem cell biology are highlighted, and the current limitations of human naïve pluripotent stem cells considered, particularly in the context of differentiation.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Epigenomics , Humans
13.
Elife ; 62017 03 23.
Article in English | MEDLINE | ID: mdl-28332981

ABSTRACT

Long-range cis-regulatory elements such as enhancers coordinate cell-specific transcriptional programmes by engaging in DNA looping interactions with target promoters. Deciphering the interplay between the promoter connectivity and activity of cis-regulatory elements during lineage commitment is crucial for understanding developmental transcriptional control. Here, we use Promoter Capture Hi-C to generate a high-resolution atlas of chromosomal interactions involving ~22,000 gene promoters in human pluripotent and lineage-committed cells, identifying putative target genes for known and predicted enhancer elements. We reveal extensive dynamics of cis-regulatory contacts upon lineage commitment, including the acquisition and loss of promoter interactions. This spatial rewiring occurs preferentially with predicted changes in the activity of cis-regulatory elements and is associated with changes in target gene expression. Our results provide a global and integrated view of promoter interactome dynamics during lineage commitment of human pluripotent cells.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Human Embryonic Stem Cells/physiology , Humans , Promoter Regions, Genetic , Regulatory Elements, Transcriptional , Transcription, Genetic
14.
Cell Stem Cell ; 20(6): 874-890.e7, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28343983

ABSTRACT

Human pluripotent stem cells (PSCs) exist in naive and primed states and provide important models to investigate the earliest stages of human development. Naive cells can be obtained through primed-to-naive resetting, but there are no reliable methods to prospectively isolate unmodified naive cells during this process. Here we report comprehensive profiling of cell surface proteins by flow cytometry in naive and primed human PSCs. Several naive-specific, but not primed-specific, proteins were also expressed by pluripotent cells in the human preimplantation embryo. The upregulation of naive-specific cell surface proteins during primed-to-naive resetting enabled the isolation and characterization of live naive cells and intermediate cell populations. This analysis revealed distinct transcriptional and X chromosome inactivation changes associated with the early and late stages of naive cell formation. Thus, identification of state-specific proteins provides a robust set of molecular markers to define the human PSC state and allows new insights into the molecular events leading to naive cell resetting.


Subject(s)
Antigens, Differentiation/biosynthesis , Gene Expression Profiling , Gene Expression Regulation/physiology , Membrane Proteins/biosynthesis , Pluripotent Stem Cells/metabolism , Cell Line , Humans , Pluripotent Stem Cells/cytology
15.
Cell Stem Cell ; 20(1): 102-111, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27989768

ABSTRACT

Sex chromosome dosage compensation is essential in most metazoans, but the developmental timing and underlying mechanisms vary significantly, even among placental mammals. Here we identify human-specific mechanisms regulating X chromosome activity in early embryonic development. Single-cell RNA sequencing and imaging revealed co-activation and accumulation of the long noncoding RNAs (lncRNAs) XACT and XIST on active X chromosomes in both early human pre-implantation embryos and naive human embryonic stem cells. In these contexts, the XIST RNA adopts an unusual, highly dispersed organization, which may explain why it does not trigger X chromosome inactivation at this stage. Functional studies in transgenic mouse cells show that XACT influences XIST accumulation in cis. Our findings therefore suggest a mechanism involving antagonistic activity of XIST and XACT in controlling X chromosome activity in early human embryos, and they highlight the contribution of rapidly evolving lncRNAs to species-specific developmental mechanisms.


Subject(s)
Chromosomes, Human, X/genetics , Embryonic Development/genetics , RNA, Long Noncoding/genetics , RNA, Untranslated/genetics , Animals , Blastocyst/metabolism , Databases as Topic , Human Embryonic Stem Cells/metabolism , Humans , Mice , Pluripotent Stem Cells/metabolism , RNA, Long Noncoding/metabolism , RNA, Untranslated/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Transgenes
16.
Cell Rep ; 17(10): 2700-2714, 2016 12 06.
Article in English | MEDLINE | ID: mdl-27926872

ABSTRACT

Through the histone methyltransferase EZH2, the Polycomb complex PRC2 mediates H3K27me3 and is associated with transcriptional repression. PRC2 regulates cell-fate decisions in model organisms; however, its role in regulating cell differentiation during human embryogenesis is unknown. Here, we report the characterization of EZH2-deficient human embryonic stem cells (hESCs). H3K27me3 was lost upon EZH2 deletion, identifying an essential requirement for EZH2 in methylating H3K27 in hESCs, in contrast to its non-essential role in mouse ESCs. Developmental regulators were derepressed in EZH2-deficient hESCs, and single-cell analysis revealed an unexpected acquisition of lineage-restricted transcriptional programs. EZH2-deficient hESCs show strongly reduced self-renewal and proliferation, thereby identifying a more severe phenotype compared to mouse ESCs. EZH2-deficient hESCs can initiate differentiation toward developmental lineages; however, they cannot fully differentiate into mature specialized tissues. Thus, EZH2 is required for stable ESC self-renewal, regulation of transcriptional programs, and for late-stage differentiation in this model of early human development.


Subject(s)
Cell Differentiation/genetics , Cell Self Renewal/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Human Embryonic Stem Cells/metabolism , Animals , Cell Proliferation/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Polycomb Repressive Complex 2/genetics , Polycomb-Group Proteins/genetics , Sequence Deletion , Single-Cell Analysis
17.
Dev Cell ; 39(1): 104-115, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27728778

ABSTRACT

Primordial germ cell (PGC) development is characterized by global epigenetic remodeling, which resets genomic potential and establishes an epigenetic ground state. Here we recapitulate PGC specification in vitro from naive embryonic stem cells and characterize the early events of epigenetic reprogramming during the formation of the human and mouse germline. Following rapid de novo DNA methylation during priming to epiblast-like cells, methylation is globally erased in PGC-like cells. Repressive chromatin marks (H3K9me2/3) and transposable elements are enriched at demethylation-resistant regions, while active chromatin marks (H3K4me3 or H3K27ac) are more prominent at regions that demethylate faster. The dynamics of specification and epigenetic reprogramming show species-specific differences, in particular markedly slower reprogramming kinetics in the human germline. Differences in developmental kinetics may be explained by differential regulation of epigenetic modifiers. Our work establishes a robust and faithful experimental system of the early events of epigenetic reprogramming and regulation in the germline.


Subject(s)
Body Patterning/genetics , DNA Methylation/genetics , Germ Cells/metabolism , Animals , DNA Transposable Elements/genetics , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...